Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
J Thromb Haemost ; 16(5): 842-848, 2018 05.
Article in English | MEDLINE | ID: mdl-29532628

ABSTRACT

Essentials Direct oral anticoagulants (DOACs) do not require laboratory monitoring currently. DOAC specific measurements were performed at trough in patients with atrial fibrillation. Patients who developed thromboembolic events showed lower DOAC plasma levels. This study supports the concept of measuring DOAC levels at steady state. SUMMARY: Background Direct oral anticoagulants (DOACs) are administered at fixed doses without the need for dose adjustment according to laboratory testing. High interindividual variability in drug blood levels has been shown with all DOACs. To evaluate a possible relationship between DOAC C-trough anticoagulant levels and thromboembolic events, 565 consecutive naive patients with atrial fibrillation (AF) were enrolled in this study performed within the START Laboratory Registry. Methods DOAC-specific measurements (diluted thrombin time or anti-activated factor II calibrated for dabigatran; anti-activated FX calibrated for rivaroxaban or apixaban) at C-trough were performed locally at steady state within 15-25 days after the start of treatment. For each DOAC, the interval of C-trough levels, from the limit of quantification to the highest value, was subdivided into four equal classes, and results were attributed to these classes; the median values of results were also calculated. Thromboembolic complications occurring during 1 year of follow-up were recorded. Results Thromboembolic events (1.8%) occurred in 10 patients who had baseline C-trough levels in the lowest class of drug levels. The incidence of thromboembolic events among patients with DOAC C-trough levels in the lowest level class was 2.4%, and that in the remaining groups was 0%. The patients with thrombotic complications also had a higher mean CHA2 DS2 -VASc score than that of the total patient population: 5.3 (95% confidence interval [CI] 4.3-6.3 versus 3.0 (95% CI 2.9-3.1). Conclusion In this study cohort, thrombotic complications occurred only in DOAC-treated AF patients who had very low C-trough levels, with a relatively high CHA2 DS2 -VASc score. Larger studies are warranted to confirm these preliminary observations.


Subject(s)
Antithrombins/administration & dosage , Antithrombins/blood , Atrial Fibrillation/drug therapy , Drug Monitoring/methods , Factor Xa Inhibitors/administration & dosage , Factor Xa Inhibitors/blood , Thromboembolism/prevention & control , Administration, Oral , Adult , Aged , Aged, 80 and over , Antithrombins/adverse effects , Atrial Fibrillation/blood , Atrial Fibrillation/complications , Atrial Fibrillation/diagnosis , Blood Coagulation Tests , Dabigatran/administration & dosage , Dabigatran/adverse effects , Dabigatran/blood , Factor Xa Inhibitors/adverse effects , Female , Humans , Male , Middle Aged , Predictive Value of Tests , Preliminary Data , Pyrazoles/administration & dosage , Pyrazoles/adverse effects , Pyrazoles/blood , Pyridones/administration & dosage , Pyridones/adverse effects , Pyridones/blood , Registries , Risk Assessment , Risk Factors , Rivaroxaban/administration & dosage , Rivaroxaban/adverse effects , Rivaroxaban/blood , Thromboembolism/blood , Thromboembolism/diagnosis , Thromboembolism/etiology , Time Factors , Treatment Outcome
2.
Mol Imaging Biol ; 17(5): 697-703, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25761907

ABSTRACT

PURPOSE: Binding of trastuzumab to HER2 receptors can be impaired by steric hindrance caused by mucin MUC4. As mucolytic drugs can breakdown disulfide bonds of mucoproteins, we checked if this approach could positively affect zirconium-89-labeled trastuzumab ([(89)Zr]T) binding/uptake. PROCEDURES: The effect of N-acetylcysteine (NAC) and MUC4 knockdown/stimulation on [(89)Zr]T binding/uptake were evaluated in MCF7(HER2-), BT474 and SKBr3(HER2+/MUC4-), and JIMT1(HER2+/MUC4+) cell lines. The results were then validated in SKBR3 and JIMT1 tumor-bearing nude mice with a microPET-CT and ex vivo analysis. RESULTS: Significant increases in [(89)Zr]T binding/uptake were observed in JIMT1 cells following MUC4 knockdown (62.4 ± 6.5%) and exposure to NAC (62.8 ± 19.4%). Compared to controls, mice treated with NAC showed a significant increase in [(89)Zr]T uptake in MUC4 tumors on microPET-CT (SUVmean (18.3 ± 4.7%), SUVmax (41.7 ± 8.4%)) and individual organ counting (37.3 ± 18.3%). In contrast, no significant differences were observed in SKBr3. CONCLUSION: NAC can enhance [(89)Zr]T accumulation and improve the HER2 imaging of MUC4-overexpressing tumors. The potential positive impact on trastuzumab-based treatment deserves further investigation.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacokinetics , Expectorants/pharmacology , Mammary Neoplasms, Experimental/pathology , Molecular Imaging/methods , Mucins/drug effects , Receptor, ErbB-2/metabolism , Acetylcysteine , Animals , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Female , Humans , Mammary Neoplasms, Experimental/diagnostic imaging , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Nude , Mice, Transgenic , Mucin-4/genetics , Mucin-4/metabolism , Positron-Emission Tomography/methods , Tissue Distribution , Xenograft Model Antitumor Assays
3.
Int J Lab Hematol ; 37(1): 71-8, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24731219

ABSTRACT

INTRODUCTION: Although most guidelines for quality assessment of INR PMs recommend specific procedures, no clear regulation or methodology is required for outpatients in our country. We have developed a specific INR portable monitor (PM) quality control system within our telemedicine organization to check over time quality performances and plan corrective actions. METHODS: Based on current guidelines for laboratory QC, the following aspects were assessed: suitability of PM, defined in terms of imprecision and accuracy; intra-assay imprecision, defined according to monthly revision of Levey-Jennings cards with data from each peripheral healthcare unit (PHU), using an internal QC provided by the manufacturer (CV ± 20% considered as acceptable); quarterly accuracy study, for assessing agreement between analytical instruments, based on duplicate analysis of three samples with INR values reflecting different therapeutic ranges (differences ± 0.5 considered as acceptable); external quality assessment (NEQAS). RESULTS: In the nine PHU, 18 portable monitors were used to perform 22 929 test during year 2010. Analytical imprecision was low, showing CVs always <5%. Accuracy check showed two of 216 results out of range (0.92%), thus providing timely indication for instrument replacement. The external QC NEQAS showed optimal performance. CONCLUSION: The current protocol for INR PMs quality assessment was effective to establish and maintain a reliable control of devices, ensuring the quality of analytical data over time. National authorities should be prompted to guarantee and apply correct protocols for INR-PM use.


Subject(s)
International Normalized Ratio/instrumentation , International Normalized Ratio/standards , Prothrombin Time/instrumentation , Prothrombin Time/standards , 4-Hydroxycoumarins/pharmacokinetics , 4-Hydroxycoumarins/therapeutic use , Adult , Aged , Aged, 80 and over , Drug Monitoring/instrumentation , Drug Monitoring/methods , Drug Monitoring/standards , Female , Humans , Indenes/pharmacokinetics , Indenes/therapeutic use , Male , Middle Aged , Point-of-Care Systems/standards , Quality Control , Reproducibility of Results , Vitamin K/antagonists & inhibitors , Vitamin K/pharmacokinetics , Vitamin K/therapeutic use , Young Adult
4.
Contrast Media Mol Imaging ; 9(2): 154-60, 2014.
Article in English | MEDLINE | ID: mdl-24523060

ABSTRACT

Melanoma is the most dangerous form of skin cancer and its incidence is rising each year. Because the current methods of diagnosis based on the visual aspect of the tumor show limitations, several new techniques are emerging to help in this diagnosis, amongst which are magnetic resonance imaging (MRI) and electron paramagnetic resonance (EPR). The origin of the typical contrast pattern observable in melanoma in T1 - and T2 -weighted images remains to be elucidated and is a source of controversy. In addition, melanin could create sufficient magnetic inhomogeneities to allow its visualization on T2 *-weighted images using high-field MRI. In order to elucidate the possible role of melanin in the MRI contrast of melanoma, the present study was designed to correlate the paramagnetic content in melanin pigment to the contrast on T1 -, T2 - and T2 *-weighted images. MR images were obtained in vivo at 11.7 T using four types of experimental tumors with different pigmentations (B16, HBL, LND1 melanomas and KHT sarcomas). The paramagnetic content in melanin pigment was measured by EPR. No significant correlation was observed between the content in melanin and the relaxation times T1 , T2 and T2 *, emphasizing that the presence of pigment alone has negligible effect on the MRI contrast.


Subject(s)
Electron Spin Resonance Spectroscopy/methods , Magnetic Resonance Imaging/methods , Melanins/chemistry , Melanoma, Experimental/diagnosis , Animals , Contrast Media/chemistry , Humans , Melanoma, Experimental/pathology , Mice
6.
Pigment Cell Res ; 17(3): 275-80, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15140073

ABSTRACT

Glutathione (GSH) and its precursor cysteine (Cys) are both known to react within any cells with oxidative species and thus play an important role in cellular defense mechanisms against oxidative stress. In melanocytes, these are also important precursors of melanogenesis by reacting non-enzymatically with l-dopaquinone to form the sulfur-containing pheomelanin. Our aim was to assess pigment role in the cellular radioprotection mechanism using a human melanoma cell model of mixed-type melanin under GSH depletion to obtain a radiosensitizing effect. The latter has been achieved either by Cys deprivation or GSH specific depletion. We first compared cell survival of Cys-deprived and GSH-depleted cells vs. control cells. Cys deprivation was achieved by decreasing Cys concentration in the culture medium for 24 h. In this condition, no toxicity was observed, Cys and GSH levels decreased, melanogenesis switched to a higher eumelanin synthesis and cells were significantly more resistant to 10-Gy dose of ionizing radiations than untreated cells. Glutathione depletion was achieved with the gamma-glutamylcysteine synthetase inhibitor buthionine-S-sulfoximine (BSO) for 24 h at 50 microM, a concentration yielding no toxicity. In this condition, intracellular GSH level decreased but no change in pigmentation was observed and cells were slightly but significantly more sensitive to radiation than the control. We then compared DNA radio-induced damages by Comet assay in control cells, cells treated as above and cells with stimulated pigmentation by increasing Tyr concentration in the medium. Our results showed that, when intracellular eumelanin content increased, DNA damage decreased. By contrast, DNA damage increased in cells treated with BSO alone. It is concluded that increasing the intracellular eumelanin content by the melanin precursor Tyr or by favoring the Pheo- to Eumelanin switch, compensates for the loss of the two intracellular radioprotectors that are GSH and Cys.


Subject(s)
Cysteine/physiology , Glutathione/physiology , Melanoma/radiotherapy , Radiation Tolerance/physiology , Buthionine Sulfoximine/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Comet Assay , DNA Damage , Dose-Response Relationship, Radiation , Humans , Melanoma/drug therapy , Melanoma/pathology , Pigmentation , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/pharmacology
7.
Br J Cancer ; 89(10): 2004-15, 2003 Nov 17.
Article in English | MEDLINE | ID: mdl-14612916

ABSTRACT

Alpha-melanocyte stimulating hormone (alpha-MSH) is known to have pleiotrophic functions including pigmentary, anti-inflammatory, antipyretic and immunoregulatory roles in the mammalian body. It is also reported to influence melanoma invasion with levels of alpha-, beta- and gamma-MSH correlated clinically with malignant melanoma development, but other studies suggest alpha-MSH acts to retard invasion. In the present study, we investigated the action of alpha-MSH on three human melanoma cell lines (HBL, A375-SM and C8161) differing in metastatic potential. alpha-melanocyte-simulating hormone reduced invasion through fibronectin and also through a human reconstructed skin composite model for the HBL line, and inhibited proinflammatory cytokine-stimulated activation of the NF-kappaB transcription factor. However, A375-SM and C8161 cells did not respond to alpha-MSH. Immunofluorescent microscopy and Western blotting identified melanocortin-1 receptor (MC-1R) expression for all three lines and MC-2R on HBL and A375-SM lines. Receptor binding identified a similar affinity for alpha-MSH for all three lines with the highest number of binding sites on HBL cells. Only the HBL melanoma line demonstrated a detectable cyclic adenosine monophosphate (cAMP) response to alpha-MSH, although all three lines responded to acute alpha-MSH addition (+(-)-N(6)-(2-phenylisopropyl)-adenosine (PIA)) with an elevation in intracellular calcium. The nonresponsive lines displayed MC-1R polymorphisms (C8161, Arg (wt) 151/Cys 151; A375-SM, homozygous Cys 151), whereas the HBL line was wild type. Stable transfection of the C8161 line with wild-type MC-1R produced cells whose invasion was significantly inhibited by alpha-MSH. From this data, we conclude that alpha-MSH can reduce melanoma cell invasion and protect cells against proinflammatory cytokine attack in cells with the wild-type receptor (HBL).


Subject(s)
Melanoma/pathology , Neoplasm Invasiveness , Skin Neoplasms/pathology , alpha-MSH/pharmacology , Cytokines/pharmacology , Humans , Inflammation , Keratinocytes , Neoplasm Metastasis , Tumor Cells, Cultured
8.
Int J Cancer ; 94(4): 586-90, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11745448

ABSTRACT

The aim of the present work was to investigate the origin and half-life of endogenous S100B protein reported by many investigators as a useful melanoma serum marker. Within cells, S100B protein exists in homo- or heterodimer form containing mainly Ca(++), having a substantial fraction bound to membranes. As such, S100B is believed to be involved in the regulation of cytoskeleton. Also, a role in the cell cycle progression has been suggested. Although S100B appears having important intracellular functions, proofs of its secretion, at least at concentrations such as the ones measured in melanoma patients, are still lacking. Consistent with this view is the fact that immunohistology for S100 protein reported by numerous authors clearly indicate an exclusive intracellular staining. For these reasons, it was of a major interest to investigate how and when S100B is shed to the blood. Knowing that significant S100B levels are seen only in stage IV patients, we hypothesized that cell death may be the major source of circulating S100B protein in these patients. This hypothesis was studied in an in vitro model simulating cell death and in vivo in melanoma and other cancer patients undergoing highly cytotoxic regional immunochemotherapy using isolated limb perfusion with tumor necrosis factor and melphalan, as well as in tumor exudates and pleural fluids. Our results strongly suggest melanoma and endothelial cell death and subsequent continuous drainage to the blood as the major mechanism behind S100B release to the blood circulation. We estimated the endogenous S100B protein half-life to be about 30 min.


Subject(s)
Calcium-Binding Proteins/blood , Calcium-Binding Proteins/metabolism , Melanoma/blood , Nerve Growth Factors/blood , Nerve Growth Factors/metabolism , S100 Proteins , Sarcoma/blood , Biomarkers, Tumor , Calcium/metabolism , Cell Death , Dimerization , Endothelium, Vascular/cytology , Hemangiosarcoma/metabolism , Humans , Kinetics , Melphalan/metabolism , Necrosis , Perfusion , S100 Calcium Binding Protein beta Subunit , Time Factors , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/metabolism , Umbilical Veins/metabolism
10.
J Immunoassay Immunochem ; 22(4): 299-321, 2001.
Article in English | MEDLINE | ID: mdl-11816800

ABSTRACT

CELISA, or cellular enzyme-linked immunosorbent assay, is a powerful and easy to use technique to study cell surface antigens under different stimulations. Nevertheless, some factors must be discussed and optimized prior to reaching a reproducible CELISA. These include the choice of cell density, fixative agent, blocking agent, culture medium, optimal antibody dilutions, and incubation time. In this paper, we first present a short review of some references devoted to CELISA by means of a comparison of these parameters, followed by their description. Then, we describe and study these different parameters using practical examples comparing TNF-induced ICAM-1 expression as an end point, on HBL melanoma and HUVEC. These cell lines were also chosen because they differ in their ability to grow as discontinuous and continuous layers, respectively. Furthermore, we designed a comprehensive flow chart, as well as a complete step-by-step protocol for CELISA optimization.


Subject(s)
Antigens, Surface/analysis , Enzyme-Linked Immunosorbent Assay/methods , Buffers , Cell Adhesion , Cell Count , Cell Line, Tumor , Cells, Cultured , E-Selectin/analysis , E-Selectin/immunology , Humans , Immunoenzyme Techniques , Intercellular Adhesion Molecule-1/analysis , Intercellular Adhesion Molecule-1/immunology , Mycoplasma/physiology , Time Factors , Trypsin/pharmacology
11.
Radiat Res ; 154(5): 497-502, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11025646

ABSTRACT

The relationship between cell pigmentation and radiosensitivity was investigated in two selected human melanoma cell lines with different melanin content (mixed type: eumelanin and pheomelanin, and pheomelanotic phenotypes). The same study was also done after stimulation of melanogenesis (1) by addition of the melanin precursor l-tyrosine to each of the cell lines separately and (2) by irradiation alone with doses ranging from 0 to 10 Gy. We found that a decrease in cell radiosensitivity was correlated with the type of melanin, with a clear involvement of eumelanin rather than pheomelanin. Increasing the intracellular content of both melanins promoted the growth of irradiated cells. Moreover, at a dose of 10 Gy, both tyrosinase activity and melanin cell content were significantly increased in the absence of any other melanogenesis promoter. Our data suggest that the amount of intracellular melanin is inversely related to the radiosensitivity of melanoma cells and may explain at least in part the controversial responses to ionizing radiations reported for melanoma.


Subject(s)
Melanins/physiology , Melanoma/radiotherapy , Radiation Tolerance/physiology , Cell Survival/physiology , Cell Survival/radiation effects , Dose-Response Relationship, Radiation , Humans , Melanins/metabolism , Melanoma/pathology , Pigmentation , Tumor Cells, Cultured
12.
Pigment Cell Res ; 13 Suppl 8: 68-72, 2000.
Article in English | MEDLINE | ID: mdl-11041360

ABSTRACT

The invasion of melanoma is complex and multi-staged and involves changes in both cell/extracellular matrix (ECM) and cell/cell interactions. Female steroids and alpha-MSH have also been reported to influence metastatic melanoma progression, but their mechanisms of action are unknown. Accordingly, our aim was to establish in vitro models to examine (a) the influence of sex steroids and alpha-melanocyte-stimulating hormone (alpha-MSH) on tumour invasion and the influence of (b) ECM proteins and (c) adjacent cells on melanoma invasion. In the first model, melanoma cell invasion through fibronectin over 20 hr under serum-free conditions was used to investigate the effects of 17beta-oestradiol and oestrone on the invasion of human melanoma cell lines, A375-SM and HBL. A375-SM, but not HBL cells, proved very susceptible to inhibition by female steroids. However, invasion of the HBL line was inhibited by alpha-MSH. Using the second model of reconstructed human skin based on de-epidermised acellular dermis, we found that the HBL cells on their own failed to invade into the dermis (irrespective of the presence or absence of the basement membrane). However, there was a significant synergistic interaction between keratinocytes, fibroblasts and HBL cells, such that a modest invasion of HBLs into the dermis was seen within 2 weeks when other skin cells were present. In contrast, A375-SM cells showed a significant ability to invade the dermis in the absence of other cells, with less invasion when other skin cells were present. In summary, these models have provided new information on the extent to which melanoma cell invasion is sensitive to oestrogenic steroids and to alpha-MSH and to interaction, not only with adjacent skin cells but also to the presence of basement membrane antigens.


Subject(s)
Estradiol/metabolism , Estrone/metabolism , Melanoma/physiopathology , Estradiol/pharmacology , Estrone/pharmacology , Female , Humans , Neoplasm Invasiveness , Skin/pathology , Tumor Cells, Cultured , alpha-MSH/metabolism , alpha-MSH/pharmacology
13.
Melanoma Res ; 10(4): 323-30, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10985666

ABSTRACT

This study was undertaken to investigate whether alpha-melanocyte stimulating hormone (alphaMSH) influences the interaction of melanoma cells with T-lymphocytes in the light of previous work from our laboratories showing that alphaMSH can reduce tumour necrosis factor-alpha (TNFalpha) stimulated ICAM-1 upregulation in both normal and transformed melanocytes. Two cutaneous melanoma cell lines--A375-SM and HBL--were examined initially. A375-SM cells gave only a two-fold increase in T-cell proliferation, which was not much improved by the pretreatment of the melanoma cells with cytokines. HBL cells induced a three-fold increase in T-cell proliferation, which was slightly enhanced by the addition of cytokines. Neither cell line expressed B7(1), HBL cells expressed a low level of B7(2), whereas A375-SM cells had little, if any, B7(2) expression. Addition of alphaMSH reduced the interaction between these cutaneous melanoma cells and T-lymphocytes in some, but not all, conditions. An ocular melanoma cell line transfected with B7 showed a modest interaction with T-cells (in two out of three donors) and this response was reduced by the addition of alphaMSH. Pretreatment of the transfected line with cytokines markedly enhanced stimulation of T-cell proliferation by these tumour cells, and alphaMSH reduced the interaction between melanoma cells and T-cells for two out of three donors. In summary, under experimental conditions where melanoma cell stimulation of T-cells occurred (generally pretreatment of the cells with interferon-gamma gave the most convincing response), alphaMSH reduced this response in the majority of experiments, providing preliminary evidence to confirm the hypothesis that MSH may assist melanoma cells to evade interaction with immune cells.


Subject(s)
Cell Communication/drug effects , Eye Neoplasms/metabolism , Melanoma/metabolism , Skin Neoplasms/metabolism , T-Lymphocytes/metabolism , alpha-MSH/pharmacology , Antibodies, Monoclonal , Antigens, CD/metabolism , B7-1 Antigen/metabolism , B7-2 Antigen , CD58 Antigens/metabolism , Coculture Techniques , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class II/metabolism , Humans , Intercellular Adhesion Molecule-1/metabolism , Lymphocyte Activation , Membrane Glycoproteins/metabolism , Transfection , Tumor Cells, Cultured/metabolism
14.
J Biol Chem ; 275(21): 15629-36, 2000 May 26.
Article in English | MEDLINE | ID: mdl-10821844

ABSTRACT

We have previously shown that alpha-melanocyte-stimulating hormone (alpha-MSH) can oppose tumor necrosis factor alpha activation of NF-kappaB (1-2 h) and intercellular adhesion molecule 1 up-regulation (mRNA by 3 h and protein by 24 h) in melanocytes and melanoma cells. The present study reports on the ability of four MSH peptides to control intracellular peroxide levels and glutathione peroxidase (GPx) activity in pigmentary and nonpigmentary cells. In human HBL melanoma and HaCaT keratinocytes tumor necrosis factor alpha and H(2)O(2) both activated GPx in a time- and concentration-dependent manner (by 30-45 min). alpha-MSH peptides were found to inhibit the stimulated GPx activity and had biphasic dose-response curves. MSH 1-13 and MSH [Nle(4)-d-Phe(7)] achieved maximum inhibition at 10(-10) and 10(-12) m, respectively. Higher concentrations (10-100 fold) of MSH 4-10 and MSH 11-13 were required to produce equivalent levels of inhibition. alpha-MSH was also capable of reducing peroxide accumulation within 15 min, and again this inhibition was biphasic. The data support a role of alpha-MSH in acute protection of cells to oxidative/cytokine action that precedes NF-kappaB and GPx activation. The rapidity and potency of the response to alpha-MSH in pigmentary and nonpigmentary cells suggest this to be a central role of this peptide in cutaneous cells.


Subject(s)
Cytokines/pharmacology , Glutathione Peroxidase/metabolism , Oxidative Stress/drug effects , Peroxides/pharmacology , alpha-MSH/pharmacology , 1-Methyl-3-isobutylxanthine/pharmacology , Colforsin/pharmacology , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Humans , Hydrogen Peroxide/pharmacology , Keratinocytes , Melanoma , NF-kappa B/metabolism , Peptide Fragments/pharmacology , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/pharmacology
15.
Br J Dermatol ; 142(2): 210-22, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10730751

ABSTRACT

The purpose of this study was to compare the invasive properties of normal human cutaneous melanocytes and of a cutaneous melanoma cell line (HBL) in a three-dimensional model of reconstructed human skin. Specifically, we asked to what extent the pigmentary and invasive behaviour of both cells is influenced by their interaction with adjacent skin cells (keratinocytes and fibroblasts) and the basement membrane (BM). In the presence of a BM, normal human melanocytes within this model remained within the basal layer of keratinocytes and did not pigment spontaneously. When the BM was removed, melanocytes were found suprabasally and pigmented extensively. No significant invasion of melanocytes into the dermis was detected in the presence or absence of the BM. HBL melanoma cells showed no significant ability to invade into the dermis in the absence of other cells, irrespective of the presence or absence of the BM. However, when added to keratinocytes and fibroblasts, HBL cells showed a capacity to invade into the dermis, both in the presence and absence of the BM. Associated with HBL invasion into the dermis, we noted significant keratinocyte entry into the dermis. On their own, keratinocytes entered the dermis in the absence of the BM but showed no significant penetration into the dermis when the BM was present. In summary, this model demonstrates clear differences between melanocytes and a melanoma cell line with respect to their invasive properties. It also allows demonstration of interactions between cells, and between cells and the BM. The study also provides evidence for a synergistic interaction between this melanoma cell line and keratinocytes in penetrating the BM.


Subject(s)
Melanoma/pathology , Models, Biological , Skin Neoplasms/pathology , Basement Membrane/pathology , Cell Communication , Fibroblasts/pathology , Humans , In Vitro Techniques , Keratinocytes/pathology , Melanocytes/pathology , Neoplasm Invasiveness , Tumor Cells, Cultured
16.
J Invest Dermatol ; 113(4): 560-6, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10504441

ABSTRACT

Alpha-melanocyte-stimulating hormone is produced by several different cell types including neural cells, endothelial cells, monocytes, and keratinocytes. A biologic role in melanocyte pigmentation is widely recognized, but more recent studies describe a part in modulating inflammatory and immune responses. The aim of the this study was to investigate the mechanism by which alpha-melanocyte-stimulating hormone antagonizes proinflammatory cytokine action. We report that alpha-melanocyte-stimulating hormone (10-9 M) was effective in opposing a tumor necrosis factor-alpha stimulated increase in NF-kappaB DNA binding activity in: (i) normal ocular melanocytes; (ii) cells cultured from ocular melanoma tumors; and (iii) two cutaneous melanoma cell lines. NF-kappaB is activated by many inflammatory mediators and controls transcription of genes required for immune and inflammatory responses. The transcription factor complex was positively identified as the p50/p65 heterodimer, recognized to have transcriptional activating potential. Maximum reduction of NF-kappaB DNA binding activity with alpha-melanocyte-stimulating hormone was detected 2 h after cellular stimulation and varied from between 53% and 18% depending on cell type. Whereas the acute inhibitory effects could be mimicked by elevating cyclic adenosine monophosphate, alpha-melanocyte-stimulating hormone was not found to have any effect on the relative level of IkappaBalpha protein expression over 24 h. These data show that alpha-melanocyte-stimulating hormone has a pronounced effect on NF-kappaB activity in melanocytes and melanoma cells, identifying a specific dimeric complex, and suggest this to be a key pathway by which immunomodulation/anti-inflammation may operate. The results may also be considered in the broader context of general inflammatory pathologies concerning cells which express alpha-melanocyte-stimulating hormone receptors and utilize the NF-kappaB signaling pathway.


Subject(s)
Melanocytes/immunology , Melanoma/immunology , NF-kappa B/antagonists & inhibitors , alpha-MSH/pharmacology , Cells, Cultured , Humans , NF-kappa B/physiology , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/pharmacology
18.
Cell Mol Biol (Noisy-le-grand) ; 45(7): 1053-63, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10644010

ABSTRACT

It is well known that ICAM-1 expression can be stimulated by TNF and by oxidative stress, via the activation of specific transcription factors. Two of these--NFkappaB and AP-1--can also be activated by reactive oxygen species, including the superoxide anion (also produced under TNF challenge). The latter is inactivated by superoxide dismutase of which two forms exist: Cu/Zn-SOD (cytoplasmic) and Mn-SOD (mitochondrial). We investigated whether superoxide anion direct generation or accumulation through specific SOD inhibition, may affect ICAM-1 expression in human melanoma and endothelial cells. Our results show a 20-50% increase in both SOD activities when cells were exposed to TNF or to an oxidative stress produced by Paraquat (a generator of superoxide anion radicals), both in terms of enzymes activity (zymogram) and protein levels (Western blotting and ELISA). Either with TNF or Paraquat, we could measure a significant increase of ICAM-1 expression with maxima ranging from 140 to 200%, depending on the cell line. Specific inhibition of Cu/Zn-SOD activity by DTIC (diethyldithiocarbamic acid), in presence of Paraquat or TNF, was followed by an upregulation of ICAM-1 expression (60 and 20%, respectively). In contrast, the addition of a SOD mimetic (MnTMPyP) completely inhibited Paraquat-stimulated ICAM-1 expression in melanoma cells and significantly decreased it in HUVEC (50%). In presence of TNF however, the same SOD mimetic inhibited TNF-stimulated ICAM-1 expression by 25% in melanoma and 17% in endothelial cells. In conclusion, these data provide evidence that melanoma and endothelial cells exposure to TNF or oxidative stress results in a significant increase of both Mn- and Cu/Zn-SOD activities. This increase seems to be associated with a reduction in the stimulation of ICAM-1 expression by TNF or oxidative stress.


Subject(s)
Endothelium, Vascular/metabolism , Gene Expression Regulation , Intercellular Adhesion Molecule-1/biosynthesis , Isoenzymes/physiology , Melanoma/pathology , Neoplasm Proteins/physiology , Skin Neoplasms/pathology , Superoxide Dismutase/physiology , Cells, Cultured/drug effects , Cytoplasm/enzymology , Dacarbazine/pharmacology , Endothelium, Vascular/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Intercellular Adhesion Molecule-1/genetics , Melanoma/metabolism , Metalloporphyrins/pharmacology , Mitochondria/enzymology , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Oxidative Stress , Paraquat/pharmacology , Skin Neoplasms/metabolism , Tumor Cells, Cultured/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Umbilical Veins
19.
Eur J Cancer ; 34(3): 424-6, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9640235

ABSTRACT

We have previously reported high immunoreactive alpha-MSH (IR-alpha-MSH) concentrations in melanoma patients' plasma, as well as significant amounts in melanoma metastases and cells grown in culture. Necrosis within the melanoma tumour leads to a massive proteolysis of intracellular proteins and release of cell content: this might significantly contribute to the elevated IR-alpha-MSH plasma levels measured in melanoma patients. To test this hypothesis, we studied the necrosis-related release of MSH from human melanoma cells, using a specific radioimmunoassay. The studies of fine-needle biopsies indicated that most of the human melanoma tumour exudates tested contained very high MSH concentrations (> 500 pg/ml; 14/15), while plasma levels were generally normal (< or = 25 pg/ml; 10/15). The level in an exudate from a non-melanoma tumour type was < 40 pg/ml. In vitro studies showed that release of the IR-alpha-MSH was time- and temperature-dependent, and related to cell death.


Subject(s)
Melanocyte-Stimulating Hormones/metabolism , Melanoma/metabolism , Humans , Immunohistochemistry , Melanoma/pathology , Necrosis , Radioimmunoassay , Tumor Cells, Cultured
20.
J Cell Physiol ; 175(3): 276-82, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9572472

ABSTRACT

Alpha-MSH, a proopiomelanocortin (POMC)-derived peptide, is known to be produced in the pituitary, the skin, and melanoma tumors and to possess many biological effects, mainly on melanocyte pigmentation and growth. Moreover, the melanocyte expresses adhesion molecules, including ICAM-1. The latter has been reported to play a role in melanoma spread and associated metastatic process. We conducted a study in order to evaluate the possible effect of MSH on ICAM-1 expression in human cultured malignant and normal melanocytes. Our data show that alpha-MSH inhibits ICAM-1 expression stimulated by TNF in a concentration-dependent manner, both at the protein and gene expression level. Ninety percent inhibition was obtained with 10 nM MSH, while 50% inhibition was achieved with 1 nM. Endogenous cAMP elevation with forskolin as well as an exogenous cAMP stable analogue (Sp-cAMPS) produced the same inhibitory effect. A screening of malignant melanocytes showed that inhibition of ICAM-1 expression could be achieved only in those cells expressing detectable MSH receptors and seemed to correlate with the number of binding sites. In conclusion, our data strongly suggest alpha-MSH as a potent inhibitor of ICAM-1 expression in malignant melanocytes acting through MSH receptor stimulation and subsequent cAMP increase.


Subject(s)
Intercellular Adhesion Molecule-1/biosynthesis , Melanocytes/metabolism , Melanoma/metabolism , alpha-MSH/pharmacology , Adult , Cells, Cultured , Colforsin/pharmacology , Cyclic AMP/analogs & derivatives , Cyclic AMP/biosynthesis , Cyclic AMP/pharmacology , Gene Expression Regulation, Neoplastic/physiology , Humans , Intercellular Adhesion Molecule-1/genetics , Melanoma/genetics , RNA, Messenger/analysis , Receptors, Pituitary Hormone/metabolism , Thionucleotides/pharmacology , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...